3-Hydroxy-3-Methylglutaryl-CoA Synthase 1 (HMGCS1) is a crucial enzyme in the mevalonate pathway, which plays a pivotal role in cholesterol biosynthesis and isomeroprenoid synthesis. This enzyme catalyzes the condensation of acetyl-CoA with acetoacetyl-CoA to form 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA), a precursor for mevalonate production. HMGCS1 is predominantly located in the cytoplasm and is involved in various cellular processes, including protein homodimerization and the biosynthesis of farnesyl diphosphate .
Gene Symbol: HMGCS1
Gene Name: 3-Hydroxy-3-Methylglutaryl-CoA Synthase 1
Chromosomal Location: 5p12
Protein Length: 520 amino acids
Species: Human, Mouse, Rat
Species | Gene Symbol | Chromosomal Location | Protein Length |
---|---|---|---|
Human | HMGCS1 | 5p12 | 520 |
Mouse | Hmgcs1 | 13 | 520 |
Rat | Hmgcs1 | 2q15 | 520 |
Recent studies have highlighted the oncogenic role of HMGCS1 in various cancers. It is overexpressed in several tumor types, including gastric, colon, cervical, and breast cancers, contributing to cancer progression and poor prognosis . In gastric cancer, HMGCS1 enhances tumor growth and metastasis by promoting stem cell-like phenotypes in cancer cells .
Cancer Type | HMGCS1 Expression | Clinical Implication |
---|---|---|
Gastric Cancer | Upregulated | Poor prognosis |
Colon Cancer | Overexpressed | Cancer progression |
Cervical Cancer | Overexpressed | Poor prognosis |
Breast Cancer | Overexpressed | Poor prognosis |
The crystal structure of HMGCS1 provides a basis for developing isoform-specific inhibitors. Compounds like 2-benzyl-9-(3,6-dioxocyclohexa-1,4-dienyl)-2,3,7,8-tetrahydrol-6H-pyrido[1,2-a]-pyrazine-1,4-dione have shown potential as inhibitors with a Ki below 10 micromolar . Additionally, HMGCS1 has been identified as a synthetic lethal partner of BRAF V600E, suggesting its potential as a therapeutic target in BRAF V600E-positive cancers .
Inhibitor | Ki Value | Target Specificity |
---|---|---|
2-benzyl-9-(3,6-dioxocyclohexa-1,4-dienyl)-2,3,7,8-tetrahydrol-6H-pyrido[1,2-a]-pyrazine-1,4-dione | <10 μM | HMGCS1 |
HMGCS1 is associated with several diseases, including Congenital Myopathy 3 with Rigid Spine and Alcoholic Ketoacidosis. Recent research suggests that biallelic variants in HMGCS1 may contribute to rare cases of rigid spine syndrome .
Disease | Association with HMGCS1 |
---|---|
Congenital Myopathy 3 with Rigid Spine | Genetic association |
Alcoholic Ketoacidosis | Metabolic involvement |
Rigid Spine Syndrome | Biallelic variants |
HMGCS1 catalyzes the condensation reaction between acetoacetyl-CoA (AcAc-CoA) and acetyl-CoA (Ac-CoA) to form 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA), representing the first committed step in the mevalonate pathway . This cytosolic enzyme is critical for cholesterol biosynthesis and production of isoprenoids essential for protein prenylation, affecting numerous cellular processes including signal transduction and protein trafficking.
To investigate HMGCS1 function experimentally, researchers typically employ several complementary approaches:
Gene expression modulation using RNA interference or CRISPR-Cas9 technologies to create knockdown or knockout models
Metabolic labeling with isotopically-tagged precursors to track flux through the mevalonate pathway
Pharmacological inhibition studies using pathway modulators
Recombinant protein expression for enzymatic characterization
Patient-derived cell models harboring HMGCS1 variants
The enzyme's activity influences multiple cellular pathways beyond cholesterol synthesis, including the Hedgehog signaling pathway, where cholesterol directly activates Smoothened (SMO), a G-protein coupled receptor that transmits Hedgehog signals .
HMGCS1 demonstrates tissue-specific expression patterns that inform appropriate experimental model selection. RNA-sequencing data from NCBI BioProject and GTEx portal indicate that HMGCS1 is enriched in brain and liver tissues, with lower expression levels in skeletal muscle and other tissues . This differential expression pattern is important for understanding the tissue-specific manifestations of HMGCS1-related disorders.
In myogenic lineages, HMGCS1 expression shows significant variation:
Cell Type | Expression Level (TPM) | Method |
---|---|---|
Skeletal muscle satellite cells | 235-462 | RNA-seq |
Fetal-derived skeletal muscle cells | 3-326 | RNA-seq |
Myotubes | 0-240 | RNA-seq |
Myoblasts | 31-50 | RNA-seq |
Western blotting confirms that HMGCS1 protein is more abundant in adult human skeletal muscle compared to fetal skeletal muscle . This developmental regulation may have implications for understanding the pathophysiology of HMGCS1-associated rigid spine syndrome, which primarily affects the musculoskeletal system.
To accurately assess tissue-specific expression patterns, researchers should combine multiple complementary techniques:
Quantitative PCR for sensitive detection of transcript levels
Western blotting with validated antibodies for protein quantification
Immunohistochemistry for spatial distribution analysis
Single-cell RNA sequencing for cell-type specific expression profiling
HMGCS1 and HMGCS2 are paralogues that catalyze similar biochemical reactions but function in distinct metabolic pathways and cellular compartments. Understanding these differences is essential for designing specific experimental approaches and interpreting research findings.
Feature | HMGCS1 | HMGCS2 |
---|---|---|
Cellular localization | Cytosolic | Mitochondrial |
Primary pathway | Mevalonate pathway (cholesterol synthesis) | Ketogenesis pathway |
Tissue expression | Widespread; enriched in brain and liver | Predominantly liver, with expression in colon and testis |
Associated disorder | Rigid spine syndrome | HMG-CoA synthase deficiency (OMIM# 605911) |
Genetic constraint scores | High (pLI = 1; Z = 4.12) | Low (pLI = 0; Z = 0.53) |
Despite these functional differences, the enzymes share approximately 67% sequence similarity . Notably, five of the six substituted HMGCS1 residues associated with rigid spine syndrome (Q29, C268, G297, R430, and S447) are conserved in HMGCS2 (Q66, C305, G334, R467, and S484), indicating their functional importance across both paralogues .
For experimental discrimination between the paralogues, researchers can employ:
Subcellular fractionation to isolate cytosolic versus mitochondrial compartments
Isoform-specific antibodies for Western blotting and immunoprecipitation
Gene-specific silencing approaches
Mass spectrometry with isoform-discriminating peptide analysis
Investigating the structure-function relationship of HMGCS1 provides insights into how genetic variants affect enzymatic activity and contribute to disease. Several methodological approaches can elucidate these structural features:
Protein dimerization analysis using size exclusion chromatography has shown that HMGCS1 functions as a dimer, and this quaternary structure is maintained even in disease-associated variants .
Thermal stability assessment through differential scanning fluorimetry reveals that certain mutations can compromise protein stability. For instance, three of four tested disease-associated mutants exhibited reduced thermal stability compared to wild-type HMGCS1 .
Secondary structure analysis via circular dichroism spectroscopy can detect subtle conformational changes that may affect catalytic efficiency.
Enzymatic assays measuring HMG-CoA formation rates can quantify the impact of structural alterations on catalytic function. Studies have shown that some disease-associated variants exhibit subtle changes in enzymatic activity compared to wild-type protein .
These structural investigations provide a mechanistic basis for understanding how specific amino acid substitutions in HMGCS1 lead to pathogenic outcomes. For example, functional studies of recombinant HMGCS1 with disease-associated variants (p.S447P, p.Q29L, p.M70T, p.C268S) demonstrated that while all mutants maintained dimerization, their stability and catalytic properties were differentially affected .
Rigid spine syndrome is characterized by slowly progressive or non-progressive scoliosis, neck and spine contractures, hypotonia, and respiratory insufficiency . Biallelic HMGCS1 variants identified in patients with this condition include both missense and frameshift alterations:
Patient | Family | Variant 1 | Variant 2 | Clinical Features |
---|---|---|---|---|
P1 | SPA1 | p.S447P | p.S447P | Spinal rigidity, scoliosis, respiratory insufficiency |
P2/P3 | JPN1 | p.Q29L | p.M70T | Cervical and dorso-lumbar rigidity, elevated CK |
P4 | GERM1 | p.C268S | p.G297D | Spinal rigidity, respiratory compromise |
P5 | FR1 | Frameshift | p.R430H | Scoliosis, respiratory failure with infection |
The clinical course can worsen with intercurrent disease or certain drugs in some patients . Muscle biopsies typically reveal irregularities in oxidative enzyme staining with occasional internal nuclei and rimmed vacuoles.
For researchers investigating these variants, several methodological approaches are recommended:
Comprehensive genetic analysis including segregation studies in families
In silico prediction tools to assess potential pathogenicity
Recombinant protein studies to measure effects on stability and activity
Patient-derived cell models to investigate metabolic consequences
Correlation between specific variants and clinical phenotypes
It's noteworthy that biallelic hypomorphic variants in downstream enzymes of the mevalonate pathway, including HMGCR and GGPS1, are associated with similar muscular dystrophy phenotypes, suggesting common pathogenic mechanisms .
Investigating the functional consequences of HMGCS1 mutations requires a comprehensive biochemical approach. Studies of recombinant HMGCS1 proteins carrying disease-associated variants have revealed differential effects on protein stability and catalytic function.
Thermal stability assessment using differential scanning fluorimetry has shown that three of four tested mutants (p.Q29L, p.M70T, p.C268S) exhibited reduced thermal stability compared to wild-type HMGCS1 . This decreased stability may contribute to protein dysfunction in vivo, potentially through altered protein half-life or improper folding.
Enzymatic activity measurements have detected subtle changes in catalytic function for some variants. Particularly, two of the four tested mutants showed alterations in enzymatic activity compared to wild-type protein . These changes may affect flux through the mevalonate pathway, potentially disrupting cholesterol biosynthesis and other dependent processes.
Protein dimerization analysis by size exclusion chromatography demonstrated that all tested mutants maintained their dimeric state, suggesting that quaternary structure is preserved despite the pathogenic mutations . This indicates that disease mechanisms likely involve aspects of protein function beyond simple structural disruption.
Interestingly, western blot analysis of muscle biopsy from a patient with the p.S447P variant showed comparable HMGCS1 abundance to control samples . This finding suggests that some pathogenic variants may affect function without significantly reducing protein expression or stability in vivo.
A comprehensive experimental approach for investigating HMGCS1 variants should include:
Biophysical characterization (thermal stability, CD spectroscopy)
Kinetic enzyme assays (Km, Vmax, catalytic efficiency)
Cellular studies examining pathway flux and metabolite levels
Structural analysis to map mutations to functional domains
Selecting appropriate experimental models for HMGCS1 research requires careful consideration of the specific research questions, tissue expression patterns, and technical feasibility. A multi-tiered approach often provides the most comprehensive insights.
Hepatocyte cell lines (HepG2, Huh7) are valuable for studying HMGCS1 in cholesterol metabolism given its high expression in liver tissue
Neuronal cultures can investigate brain-specific functions where HMGCS1 is also highly expressed
Myoblast/myotube culture systems are particularly relevant for investigating rigid spine syndrome mechanisms
Patient-derived fibroblasts or induced pluripotent stem cells (iPSCs) offer disease-relevant cellular contexts
Mouse models with conditional or tissue-specific Hmgcs1 knockout/knockdown
CRISPR-engineered mice carrying specific Hmgcs1 mutations that mirror human pathogenic variants
Zebrafish models for developmental studies and high-throughput screening
Purified recombinant HMGCS1 protein for in vitro enzymatic and structural studies
Cell-free expression systems for protein folding and stability assessments
When comparing in vitro and in vivo models, research has shown that cellular responses can differ significantly between cultured cells and tissue contexts . While similarities may exist at the pathway level, the specific genes altered under these pathways are often different, suggesting that underlying mechanisms of responses differ between isolated cells and complex tissues .
The choice between in vitro and in vivo approaches for HMGCS1 research presents important methodological considerations that can significantly impact experimental outcomes and interpretations.
Transcriptomic analysis comparing in vitro and in vivo responses has revealed that while certain pathway-level similarities may exist, the specific genes altered within these pathways often differ substantially between cells in culture and tissue contexts . This divergence suggests that the underlying mechanisms of cellular responses in isolated cells may not fully recapitulate the complexity of tissue environments.
For HMGCS1 specifically, several factors should guide model selection:
Pathway Context: The mevalonate pathway involves multiple enzymes and regulatory factors that may be differentially expressed or regulated in various model systems.
Tissue Interactions: HMGCS1 function in skeletal muscle may depend on interactions with other cell types (neurons, vascular cells) that are absent in monoculture systems.
Systemic Influences: Hormonal regulation and metabolic feedback mechanisms present in vivo but absent in vitro may significantly affect HMGCS1 function.
Developmental Dynamics: Studies show HMGCS1 expression varies between adult and fetal muscles , suggesting developmental regulation that may be difficult to model in vitro.
A recommended research strategy involves:
Beginning with biochemical characterization using recombinant proteins
Progressing to cellular models with appropriate tissue context
Validating key findings in animal models that recapitulate human disease features
Considering "organoid" or "tissue-on-chip" systems as intermediate complexity models
Researchers should carefully select relevant endpoints when substituting animal testing with in vitro approaches , particularly focusing on parameters that are conserved across model systems.
Accurate measurement of HMGCS1 enzymatic activity requires specific approaches that distinguish its function from related enzymes and account for the complex regulation of the mevalonate pathway.
Spectrophotometric methods: Monitoring the condensation of acetyl-CoA and acetoacetyl-CoA by measuring the decrease in absorbance at 303 nm as thioester bonds are cleaved
HPLC-based detection: Quantifying HMG-CoA formation with high sensitivity and specificity
Radiometric assays: Using 14C-labeled substrates to track product formation
Subcellular fractionation: Isolating cytosolic fractions where HMGCS1 is located, avoiding contamination from mitochondrial HMGCS2
Rapid processing: Minimizing enzyme degradation and maintaining activity
Buffer optimization: Including appropriate cofactors and stabilizing agents
Metabolic labeling: Using isotope-labeled precursors (13C-acetate) to trace carbon flux through the pathway
Mass spectrometry: Quantifying mevalonate pathway intermediates and end-products
Reporter systems: Employing transcriptional reporters responsive to mevalonate pathway activity
When working with tissue samples from patients with HMGCS1 variants, researchers should consider:
Parallel measurement of protein levels by Western blotting or mass spectrometry
Correlation of enzymatic activity with clinical severity
Comparison with other mevalonate pathway enzymes to assess pathway-wide effects
For recombinant protein studies, kinetic analysis should determine Km, Vmax, and catalytic efficiency parameters to quantify the impact of disease-associated mutations. Such approaches have revealed subtle changes in enzymatic activity for certain HMGCS1 variants associated with rigid spine syndrome .
Recombinant HMGCS1 protein provides a powerful tool for detailed biochemical characterization, enabling precise analysis of how genetic variants affect protein function. Several methodological approaches have proven effective:
Bacterial expression systems have successfully produced functional human HMGCS1, with appropriate affinity tags facilitating purification while preserving enzymatic activity . Optimizing solubility conditions is critical for obtaining properly folded, active enzyme.
Size exclusion chromatography has demonstrated that HMGCS1 functions as a dimer, and this quaternary structure is maintained even in disease-associated variants (p.S447P, p.Q29L, p.M70T, p.C268S) .
Thermal shift assays have revealed differential stability profiles between wild-type and mutant proteins. Three of four disease-associated mutants exhibited reduced thermal stability, providing mechanistic insights into protein dysfunction .
Circular dichroism spectroscopy enables detection of alterations in secondary structure elements that might contribute to functional deficits.
Enzyme activity assays measuring the condensation of acetyl-CoA and acetoacetyl-CoA have identified subtle functional changes in certain HMGCS1 variants. Two of four tested disease-associated mutants showed alterations in catalytic properties compared to wild-type enzyme .
A comprehensive approach to characterizing HMGCS1 variants includes:
Site-directed mutagenesis to introduce specific mutations
Parallel purification of wild-type and mutant proteins under identical conditions
Comparative analysis of biophysical properties (stability, structure)
Detailed kinetic characterization (Km, Vmax, substrate specificity)
Correlation of biochemical findings with clinical phenotypes
This biochemical approach has provided valuable insights into how specific amino acid substitutions in HMGCS1 contribute to rigid spine syndrome, demonstrating that disease mechanisms involve alterations in protein stability and subtle changes in enzymatic function rather than complete loss of catalytic activity .
The interconnection between HMGCS1, cholesterol biosynthesis, and Hedgehog (Hh) signaling represents a fascinating convergence of metabolic and developmental pathways with significant research implications.
HMGCS1 catalyzes a critical step in the mevalonate pathway leading to cholesterol production . Beyond its structural role in membranes, research has revealed that cholesterol functions as an instructive signaling molecule in the Hedgehog pathway, a prominent cell-cell communication system in development .
Mechanistically, cholesterol directly activates Smoothened (SMO), an orphan G-protein coupled receptor that transmits the Hedgehog signal across the membrane in all animals . Unlike many GPCRs regulated by cholesterol through their transmembrane domains, SMO is activated by cholesterol through its extracellular cysteine-rich domain (CRD) .
This relationship has several important research implications:
Dual Roles of Cholesterol: Cholesterol serves both permissive and instructive functions in Hh signaling. While depletion of cholesterol reduces cellular responses to Hh ligands, acute increases in plasma membrane cholesterol are sufficient to activate Hh signaling independently of the ligand .
Temporal Dynamics: Studies using cyclodextrin-cholesterol complexes (MβCD:cholesterol) to rapidly deliver cholesterol to cells have shown that Hh pathway activation (measured by Gli1 expression) coincides with the loading of cells with cholesterol, starting at approximately 2 hours post-treatment .
Signaling Mechanism: Cholesterol-mediated activation of the Hh pathway requires SMO activity, as demonstrated by pharmacological inhibition studies and genetic knockout experiments .
For researchers investigating this relationship, several methodological approaches are valuable:
Pharmacological modulation of HMGCS1 activity to alter cholesterol levels
Monitoring Hh pathway activation (e.g., Gli1 expression) following manipulation of cholesterol synthesis
Using fluorescent cholesterol probes to track membrane cholesterol localization
Employing SMO mutants with altered cholesterol sensitivity to dissect specific mechanisms
HMGCS1 expression exhibits dynamic patterns during development and cellular differentiation that provide important context for understanding its role in health and disease.
Transcriptomic and proteomic analyses of myogenic lineages have revealed significant variation in HMGCS1 expression across different developmental stages and cell types:
Cell/Tissue Type | HMGCS1 Expression Level | Method | Significance |
---|---|---|---|
Skeletal muscle satellite cells | High (235-462 TPM) | RNA-seq | Stem cell reserves |
Fetal skeletal muscle cells | Variable (3-326 TPM) | RNA-seq | Developmental stage |
Myotubes | Variable (0-240 TPM) | RNA-seq | Differentiated state |
Myoblasts | Moderate (31-50 TPM) | RNA-seq | Proliferative precursors |
Adult skeletal muscle | Higher than fetal | Western blot | Maturation-dependent |
Fetal skeletal muscle | Lower than adult | Western blot | Developmental regulation |
These expression patterns have several important research implications:
The enrichment of HMGCS1 in satellite cells suggests a potential role in muscle stem cell maintenance or function, which may be relevant to understanding muscle disorders associated with HMGCS1 variants .
The increased expression in adult versus fetal muscle indicates developmental regulation that may reflect changing requirements for cholesterol or isoprenoids during muscle maturation .
Similar levels of Hmgcs1 detected in mouse fast-twitch (EDL) and slow-twitch (soleus) muscles suggest that fiber-type specificity may not be a major determinant of expression .
For researchers investigating developmental regulation of HMGCS1, several methodological approaches are recommended:
Temporal expression profiling across developmental stages using qPCR and Western blotting
In situ hybridization or immunohistochemistry to visualize spatial expression patterns
Single-cell RNA sequencing to capture cell-type specific expression dynamics
Reporter constructs driven by the HMGCS1 promoter to track expression in living systems
Correlation of expression patterns with functional requirements for mevalonate pathway products
Transcriptomic methodologies offer powerful approaches for investigating HMGCS1 function in both normal physiology and disease contexts. These techniques can reveal pathway interactions, regulatory mechanisms, and downstream consequences of HMGCS1 perturbation.
RNA sequencing (RNA-seq) provides genome-wide expression data following HMGCS1 modulation, revealing both direct and indirect effects on gene expression patterns.
Targeted approaches such as NanoString technology or qPCR arrays can focus on specific pathway components with high sensitivity.
Single-cell RNA-seq enables identification of cell-specific responses to HMGCS1 alteration, particularly valuable in heterogeneous tissues like muscle.
Transcriptomic analysis has revealed that HMGCS1 perturbation affects multiple cellular processes including:
Core cellular functions (transcription, cell cycle, growth and proliferation)
Oxidative stress responses
Fibrosis pathways
These findings highlight the broad impact of mevalonate pathway disruption on cellular homeostasis.
When designing transcriptomic experiments, researchers should consider that responses may differ significantly between in vitro and in vivo systems. While similarities may exist at the pathway level, the specific genes altered within these pathways often differ between cell culture and tissue contexts .
This phenomenon has important implications for experimental design:
Multiple model systems should be employed when possible
Key findings should be validated across different experimental contexts
Pathway-level analysis may be more transferable than individual gene changes
For optimal transcriptomic investigation of HMGCS1 function:
Include appropriate time points to capture both immediate and delayed responses
Compare wild-type and HMGCS1-deficient or variant systems
Consider parallel profiling of protein and metabolite changes
Employ bioinformatic approaches that identify enriched pathways and predicted upstream regulators
Validate key findings using orthogonal techniques (qPCR, Western blotting)
These approaches can identify novel functions, regulatory mechanisms, and potential therapeutic targets related to HMGCS1 biology.
Developing therapeutic strategies targeting HMGCS1 requires understanding its role in disease pathogenesis and identifying effective intervention approaches. For HMGCS1-related disorders such as rigid spine syndrome, several therapeutic avenues warrant investigation.
Enzyme modulation strategies:
Small molecule inhibitors or activators that directly target HMGCS1
Allosteric modulators that specifically stabilize disease-associated variants
Pharmacological chaperones that improve folding and stability of variant proteins
Genetic approaches:
Gene therapy to deliver wild-type HMGCS1 to affected tissues
Antisense oligonucleotides to modulate splicing or expression
CRISPR-based strategies for correction of specific variants
Metabolic bypass strategies:
Supplementation with downstream products of the mevalonate pathway
Modulation of alternative metabolic pathways that can compensate for HMGCS1 dysfunction
Dietary interventions affecting cholesterol homeostasis
The clinical course of rigid spine syndrome associated with HMGCS1 variants can worsen with intercurrent disease or certain medications . This observation suggests that:
Preventative measures may be an important component of management
Some drugs may adversely affect pathway function and should be identified
Personalized medicine approaches based on specific variants may be necessary
When investigating potential therapeutic approaches, researchers should consider:
Tissue specificity: While HMGCS1 is widely expressed, disease manifestations primarily affect skeletal muscle, suggesting the need for muscle-targeted interventions .
Variant-specific effects: Different HMGCS1 variants exhibit varying effects on protein stability and enzymatic activity, potentially requiring tailored therapeutic approaches .
Pathway interactions: HMGCS1's role in the mevalonate pathway may allow therapeutic targeting of downstream enzymes or products as alternative approaches.
Biomarker development: Establishing reliable biomarkers of pathway activity and disease progression is essential for clinical trials and treatment monitoring.
Experience with related disorders caused by defects in downstream enzymes like HMGCR and GGPS1, which present with similar muscular dystrophy phenotypes, may inform therapeutic development for HMGCS1-related conditions .
HMGCS1 functions within the complex network of the mevalonate pathway, which involves multiple enzymes, regulatory factors, and metabolic intermediates. Understanding these interactions is essential for comprehending pathway regulation and developing targeted interventions.
HMGCS1 catalyzes the condensation of acetoacetyl-CoA and acetyl-CoA to form HMG-CoA, which serves as the substrate for HMG-CoA reductase (HMGCR) . HMGCR catalyzes the rate-limiting step in cholesterol biosynthesis, converting HMG-CoA to mevalonate. The sequential arrangement of these enzymes creates potential for:
Feedback regulation where end products (cholesterol, isoprenoids) influence upstream enzyme activities
Substrate competition between parallel pathways
Metabolic channeling where intermediates are passed directly between pathway components
Several regulatory mechanisms ensure balanced flux through the mevalonate pathway:
Transcriptional control: HMGCS1 expression is regulated by sterol regulatory element-binding proteins (SREBPs) in response to cellular sterol levels
Post-translational modifications: Phosphorylation, acetylation, and other modifications can rapidly adjust enzyme activity
Protein-protein interactions: Physical associations between pathway enzymes may form metabolic complexes for enhanced efficiency
Biallelic variants in different mevalonate pathway enzymes produce related but distinct phenotypes:
This pattern suggests common pathogenic mechanisms throughout the pathway, potentially involving disrupted prenylation of proteins important for muscle function.
To investigate HMGCS1 interactions within the mevalonate pathway, researchers can employ:
Metabolic flux analysis using isotope-labeled precursors to track carbon flow through the pathway
Protein-protein interaction studies (co-immunoprecipitation, proximity labeling) to identify physical associations
Integrative omics approaches combining transcriptomics, proteomics, and metabolomics to build comprehensive pathway models
Pharmacological perturbation using inhibitors of specific pathway enzymes to assess compensatory mechanisms
Genetic interaction screens to identify synthetic lethal or synergistic effects between pathway components
3-Hydroxy-3-Methylglutaryl-CoA Synthase 1 (HMGCS1) is a crucial enzyme in the mevalonate pathway, which is essential for the biosynthesis of cholesterol and other isoprenoids. This enzyme catalyzes the condensation of acetyl-CoA with acetoacetyl-CoA to form 3-Hydroxy-3-Methylglutaryl-CoA (HMG-CoA), a key intermediate in the pathway .
HMGCS1 plays a pivotal role in the mevalonate pathway by catalyzing the first committed step in the synthesis of HMG-CoA. This reaction involves the condensation of acetyl-CoA and acetoacetyl-CoA, which is then converted by HMG-CoA reductase (HMGCR) into mevalonate . Mevalonate is a precursor for the synthesis of cholesterol, steroid hormones, and other essential biomolecules .
Mutations or dysregulation of the HMGCS1 gene can lead to various metabolic disorders. For instance, congenital myopathy with rigid spine and cardiac sarcoidosis have been associated with abnormalities in the HMGCS1 gene . Additionally, the enzyme’s role in cholesterol biosynthesis makes it a potential target for therapeutic interventions aimed at lowering cholesterol levels .
Recombinant HMGCS1 is produced using recombinant DNA technology, which involves cloning the HMGCS1 gene into an expression vector and introducing it into a host organism, such as Escherichia coli, for protein production . The recombinant enzyme is then purified and used for various research and therapeutic applications .
Recombinant HMGCS1 is widely used in biochemical research to study the mevalonate pathway and its regulation. It is also employed in drug discovery efforts aimed at identifying inhibitors of HMGCS1 as potential cholesterol-lowering agents . Furthermore, the enzyme is used in structural biology studies to elucidate its three-dimensional structure and understand the molecular basis of its catalytic activity .